1,575
Views
7
CrossRef citations to date
0
Altmetric
Original Article

The platelet-to-lymphocyte ratio as a predictor of patient outcomes in ovarian cancer: a meta-analysis

, , , , , , & show all
Pages 448-455 | Received 01 Dec 2016, Accepted 28 Apr 2017, Published online: 01 Jun 2017

Abstract

Objectives: The platelet-to-lymphocyte ratio (PLR) is a predictive clinical biomarker for different cancers. However, the results of several studies investigating the association between the PLR and the prognosis of ovarian cancer have been inconclusive. Therefore, there is a need to conduct a meta-analysis to estimate the prognostic value of the PLR in ovarian cancer.

Methods: We searched the EMBASE, Medline, PubMed, and Web of Science databases to identify clinical studies that had evaluated the association between the PLR and ovarian cancer prognosis. Outcomes evaluated included overall survival (OS) and progression-free survival (PFS). We also analyzed PLR differences between malignant ovarian masses and the controls.

Results: Twelve relevant studies that comprised 2340 patients were selected for the meta-analysis. The results revealed that elevated PLR was significantly associated with poor OS (hazard ratio (HR) 1.63, 95% confidence interval (CI) 1.05–2.56, p < 0.01) and PFS (HR 1.61, 95% CI 1.03–2.51, p < 0.01). The PLRs in malignant cases were higher than in controls (mean difference = 63.57, 95% CI 39.47–87.66, p < 0.00001).

Conclusion: An elevated PLR is associated with poor prognosis in patients with ovarian cancer. The PLR could be employed as a prognostic marker in patients with ovarian cancer.

Introduction

Ovarian cancer (OC) is the fifth leading cause of cancer-related mortality among gynecologic cancers; however, less than 40% of women with OC were diagnosed in the early stages of the disease. A primary cytoreductive surgery followed by adjuvant chemotherapy is the standard treatment for most patientsCitation1. Even though most patients have an initial good response to chemotherapy, almost 75% of them experience a resurgence of the disease, with poor overall survivalCitation2. On account of advances in chemotherapy and surgical techniques, the prognosis of OC has improved. However, the survival of OC patients is still not optimistic. Survival statistics show 5-year survival can change according to stage and therapy; in stage 1 it was about 90%, in stage 2 it was more than 40%, in stage 3 it was almost 20%, and around 5% in stage 4Citation3. A major reason for this poor outcome is that early stages of this disease are usually asymptomatic and there are no specific diagnostic biomarkers for the patients at early disease stage.

The gold standard for differential diagnosis of adnexal masses is surgeryCitation4. Preoperatively, the standard tools for detecting OC are pelvic ultrasonography and measuring serum cancer antigen 125 levels, which could be combined with the menopausal status to calculate the risk malignancy indexCitation5. However, in daily practice, due to the performance limitations and time consumption of the standard tools, new prognostic assessment methods that are easy to use, inexpensive, and effective are needed for these patients. Research conducted in recent years has yielded many prospective biomarkers for predictors of OC prognosis, but they lack sensitivity, specificity, or reproducibility. Because of these drawbacks, they are still not accepted as part of the routine diagnostic algorithm for OC. Thus, the identification of accurate and reliable biomarkers and prognostic indicators is key for the improvement of patient outcomes and survival.

Recently, many studies have focused on hematological measurements or inflammatory factors, which are associated with the tumor microenvironment, for the diagnosis and prognosis of different cancersCitation6. Previous clinical studies have confirmed that increases in the neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio (PLR), neutrophil counts, or platelet counts were associated with negative clinical pathologic characteristics, such as a higher risk of recurrence, aggressive tumor biology, and greater disease progression, in patients with various types of cancerCitation7,Citation8. Platelets may release growth factors such as transforming growth factor β (TGF-β), platelet-derived growth factor (PDGF), and vascular endothelial growth factor (VEGF), which may contribute to the growth, progression, and metastasis of the tumor by stimulating the proliferation of ovarian tumor cellsCitation9. Furthermore, a reduced lymphocyte count is an indicator of a reduced immune response, and it is correlated with higher mortality in patients with OC than in healthy controls or patients with benign diseasesCitation10. The PLR, which is inexpensively and easily determined in routine clinical practice, was proposed to be a highly efficient and independent prognostic biomarker for many tumorsCitation7. Although there are several studies already published examining the relationship between PLR and cancer, the role of PLR in OC prognosis remains unclearCitation11.

In light of these findings, we performed a meta-analysis to determine the predictive value of the PLR for prognosis in OC, and to analyze the difference in PLR between patients with malignant ovarian masses and those with benign ones.

Methods

Search strategy

A comprehensive search strategy was employed to search the EMBASE via OvidSP, Medline via OvidSP, PubMed and Web of Science databases for all relevant literature published before March 25, 2017. The following keywords were used in accordance with the search strategy: (“ovarian neoplasms”) AND (“platelet-to-lymphocyte ratio” OR “platelet lymphocyte ratio” OR “platelet-lymphocyte ratio” OR “PLR”). The search was performed using both free text and MeSH terms. In addition, references in the related literature were manually searched to identify potentially relevant articles.

Inclusion and exclusion criteria

The full text of a selected article was further screened for eligibility, if the title and abstract met the following criteria according to either reviewer: (1) OC was diagnosed on the basis of histopathological examination; (2) the PLRs of patients were measured prior to treatment; (3) correlation of PLR with prognosis (OS and PFS) was reported; or (4) the PLR was reported in patients with benign and malignant ovarian masses. The exclusion criteria were as follows: (1) review articles, conference abstracts, editorial comments, letters, expert opinions, or case reports; (2) studies with insufficient data to perform the meta-analysis. Two reviewers independently screened the title and abstract search results.

Extraction and quality assessment of data

Two researchers collected data independently in accordance with predesigned tables, which included author, publication year, country, ethnicity, study period, sample size, disease stage, histology, treatment, cut-off value, PLR (high/low), endpoint (OS or PFS), PLR in patients with malignant ovarian masses and controls, and number of patients in each group.

The quality of the included studies was assessed independently by two investigators using the Newcastle-Ottawa Quality Assessment Scale (NOS). NOS scores >6 were considered to indicate high-quality studies. Disagreements between reviewer assessments were resolved via group discussion and consensus.

Statistical analysis

Meta-analysis was performed using RevMan5.2, which was recommended by the Cochrane Collaboration. Hazard ratios (HRs) and their 95% confidence intervals (CIs) were obtained directly from included studies for all the survival outcomes, and log (HR) and standard error were calculated accordingly; the pooled HR was calculated by the method of genetic inverse varianceCitation12. Mean differences were calculated for continuous variables. The heterogeneity among the included studies was assessed using the χ2 test, with a p-value <0.10 indicating a significant difference. The random effects model was used when heterogeneity existed, and subgroup analysis was conducted to investigate the source of the heterogeneity. A funnel plot was used to evaluate publication bias. The total effect was evaluated using the Z test, and p < 0.05 was used to identify significant effectsCitation13.

Results

Study selection and characteristics

The literature retrieval procedure is shown in . The original search algorithm retrieved 125 studies. After further discussion and consideration of the retrieved articles, 12 studies published between 2011 and 2016, involving 2340 patients, were selected for this meta-analysis. The study characteristics are summarized in . Notably, six studies were conducted in Turkey, two in China, two in Thailand, one in Poland, and one in the UK. The NOS scores, which indicated the quality of these studies, ranged from 6 to 8.

Figure 1. Flow diagram of included studies. PLR, platelet-to-lymphocyte ratio; OS, overall survival; PFS, progression-free survival.

Figure 1. Flow diagram of included studies. PLR, platelet-to-lymphocyte ratio; OS, overall survival; PFS, progression-free survival.

Table 1. Characteristics of included studies. Data for age are given as mean ± standard deviation (range).

Five of the selected eligible studies presented data regarding the association between the PLR and OSCitation14–18, five studies discussed the link between PLR and PFSCitation15–19, and six examined numerical differences in PLR between patients with malignant ovarian masses and the controlsCitation20–25. The disease characteristics in 12 studies are summarized in .

Table 2. Characteristics of disease.

PLR and OS in ovarian cancer

Five studies that comprised 1250 OC patients reported HRs for OS. The results of the analysis of these studies are displayed in Citation14–18. Our pooled results favored the low PLR patients (HR 1.63, 95% CI 1.05–2.56, p < 0.01), which indicated that a higher PLR was more strongly associated with poor OS. Heterogeneity was observed among these studies (I2 = 93%, p < 0.01). Subgroup analyses were performed to explore the source of heterogeneity; when studies were stratified by cut-off value ≤200 and cut-off value >200, the data showed that no heterogeneity was found (I2 = 0%, p = 0.76; I2 = 26%, p = 0.25, respectively) (). Subgroup analyses indicated that the cut-off value may be the source of heterogeneity.

Figure 2. Forest plots of studies evaluating hazard ratio with 95% confidence interval (CI) of platelet-to-lymphocyte ratios for overall survival.

Figure 2. Forest plots of studies evaluating hazard ratio with 95% confidence interval (CI) of platelet-to-lymphocyte ratios for overall survival.

PLR and PFS in ovarian cancer

Five studies comprising 1051 patients investigated the association between PLR and PFSCitation15–19. High PLR indicated an inferior PFS outcome, with a combined HR of 1.61 (95% CI 1.03–2.51, p < 0.01) (), which indicated that patients with a higher PLR had a higher risk of disease progression than those with a low PLR. Heterogeneity among these studies was observed (I2 = 89%, p < 0.01). When studies were subgrouped by cut-off value ≤200 and cut-off value >200, the data showed that no heterogeneity was found (I2 = 0%, p = 0.53; I2 = 56%, p = 0.13, respectively) (). Subgroup analyses indicated that the cut-off value may be the source of heterogeneity.

Figure 3. Forest plots of studies evaluating hazard ratio with 95% confidence interval (CI) of platelet-to-lymphocyte ratios for progression-free survival.

Figure 3. Forest plots of studies evaluating hazard ratio with 95% confidence interval (CI) of platelet-to-lymphocyte ratios for progression-free survival.

Numerical differences in PLR between patients with malignant and benign ovarian masses

Six studies comprising 1054 patients reported the PLR in patients with malignant ovarian masses and the controlsCitation20–25. A comparison of the malignant cases with the controls revealed a significant difference in PLR; the pooled mean difference was 63.57 (95% CI 39.47–87.66, p < 0.00001). Heterogeneity was observed (I2 = 81%, p < 0.0001). Subgroup analysis stratified the controls by benign masses and healthy tissue; the pooled data are shown in . The result indicated that the difference in controls may be the source of heterogeneity. The PLRs in the patients with malignant masses were higher than those in patients with benign masses and healthy tissue, indicating that patients with higher PLR tend to have a higher risk of OC.

Figure 4. Forest plots of studies evaluating mean differences of platelet-to-lymphocyte ratios in malignant ovarian masses and controls. 95% CI, 95% confidence interval.

Figure 4. Forest plots of studies evaluating mean differences of platelet-to-lymphocyte ratios in malignant ovarian masses and controls. 95% CI, 95% confidence interval.

Publication bias

Publication bias was evaluated by the funnel plot; indicates that potential publication bias may exist for OS and PFS. However, due to the small number of included studies, the funnel plots may not be significant.

Figure 5. Funnel plots of the meta-analysis of the impact of platelet-to-lymphocyte ratios on overall survival (A), progression-free survival (B) and mean difference between malignant ovarian masses and controls (C). SE, standard error; MD, mean difference.

Figure 5. Funnel plots of the meta-analysis of the impact of platelet-to-lymphocyte ratios on overall survival (A), progression-free survival (B) and mean difference between malignant ovarian masses and controls (C). SE, standard error; MD, mean difference.

Discussion

In this meta-analysis, by integrating the results of independent studies, elevated PLR was found to be associated with poor OS and PFS. PLRs in patients with malignant ovarian masses were higher than those in patients with benign masses. Heterogeneity was observed among the included studies; we tried to explore the source of heterogeneity by subgroup analyses. In the OS and PFS analyses, the cut-off value of PLR was the source of heterogeneity, and a higher PLR cut-off value had a higher clinical prognostic value. In the analyses of mean differences in PLR between malignant OC and controls, the control group was stratified by benign masses and healthy tissue; the difference in controls may be the source of heterogeneity.

A systematic review was conducted in 2014 to investigate the prognostic value of PLR in various cancers, with only two articles on OCCitation26. In addition, the interaction between clinical pathologic characteristics and the PLR has been illustrated in a variety of studies. Huang and colleagues reported that OC patients with a lower PLR showed a better treatment response to dose-dense chemotherapy. Thus, the PLR might be an indicator of treatment response in dose-dense chemotherapy, and can help clinicians to decide when to switch patients to another chemotherapy regimenCitation27. A study by Xiang and colleagues compared the MLR together with PLR between OC patients and controls; significant differences were observedCitation28. In a study by Kokcu and colleagues, PLR increased progressively with increases in the stage of OC, indicating that it could function as an independent prognostic factor related to the cancer stageCitation29. A study by Ashrafganjoei and colleagues reported an association between the PLR and surgical outcomes in patients with OC. High PLRs were associated with poor surgical outcomes and suboptimal debulkingCitation30. Because the results of the above studies do not match the outcome of our analysis, they have not been included in our study. However, these findings have strongly supported the prognostic value of PLR in OC.

Study limitations include the insufficient number of studies as well as their retrospective nature, which might reduce the generalizability of the results. Second, the funnel plot suggests potential publication bias. Third, several studies have evaluated PLR in combination with other biomarkers, which may be of practical value to the clinic, but the number of studies cannot support meta-analysis. In the future, well-designed and high-quality clinical studies with a larger sample size are needed to draw definitive conclusions regarding the prognostic value of PLR in OC.

The following reasons may explain why PLR could serve as an accurate and reliable prognostic marker. First, PLR can be measured in routine blood tests performed in daily clinical practice, making it easy to measure, low cost, and reproducibleCitation31,Citation32. Second, the prognostic value of the PLR has been demonstrated in a wide variety of tumor typesCitation33. However, the mechanisms by which tumors alter the PLR in patients with OC are still incompletely understood. Increases in the PLR may be linked to an increased platelet-dependent systemic inflammatory response together with a lower lymphocyte-mediated antitumor immune response, which reflect a supportive tumor microenvironment.

Platelets play major roles in hemostasis, thrombosis, inflammation, and vascular biologyCitation34. In both mouse models and patients with OC, tumors induce thrombocytosis and thrombosis by stimulating IL-6 release and hepatic thrombopoietin expressionCitation35,Citation36. Recently, a high prevalence of platelet-derived venous thromboembolism has been reported in patients with cancer. Alternatively, platelets recruited by tumor cells may act as catalysts for acceleration of tumor growth, angiogenesis, and metastasisCitation37. When the tumor volume exceeds a certain size (> 1–2 mm3), angiogenesis occurs in tumors from pre-existing blood vessels, and provides the oxygen and nutrients essential to sustain tumor growthCitation38. Experimental evidence indicates that the stimulatory effect of platelets on angiogenesis may facilitate tumor growth and metastasis. Upon activation, platelets can release a series of pro-angiogenic factors such as VEGF, PDGF, basic fibroblast growth factor (β-FGF), insulin-like growth factor 1, lysophosphatidic acid, angiopoietin, matrix metalloproteinase (MMP)-1, MMP-2, and MMP-9Citation39. These signaling ways induces angiogenesis by increasing sprouting, vessel dilation, and the detachment of pericytes from pre-existing vessels, and the subsequent increase in blood supply potently facilitates rapid tumor growth and metastasisCitation40,Citation41. These findings make the angiogenic growth factor system an attractive oncologic therapeutic targetCitation42. In addition, experimental data from several clinical trials suggest the possible benefits of anti-platelet drugs in limiting tumor initiation and progression. Many novel VEGF/VEGFR-targeted inhibitors have been reported to improve the prognosis and survival of patients more than chemotherapy alone doesCitation42, such as bevacizumab and ramucirumabCitation43–45. However, when VEGF signaling is blocked, other compensatory angiogenic signaling pathways can regulate tumor angiogenesis, including the PDGF, FGF, and hepatocyte growth factor/Met signaling pathwaysCitation46. Consequently, multi-agent therapy appears to be the optimal strategy for targeting angiogenesis. Genetic and molecular studies have led to a better understanding of biological characteristics of tumors, resulting in the development of multiple molecular targeted agents. These agents, which include sorafenib, sunitinib, axitinib, and pazopanib, aim to neutralize VEGF, PDGF, FGF, and other signaling pathwaysCitation47–50. Because OC is recognized as an extremely vascularized tumor, antiangiogenic agents have become a key focus of treatment.

In a variety of tumors, cancer cells often disrupt immune system homeostasis to their advantage in order to escape from immune surveillance. Indeed, an immune-suppressive milieu is a common in the tumor microenvironment. Appropriate immune responses, which have been intensively studied in recent decades, play an important role in controlling cancer growthCitation51,Citation52. Lymphocytes are key mediators of immune surveillance and immune editing, and lymphocyte infiltration into the tumor microenvironment is a prerequisite for an immunologic antitumor reaction. High lymphocyte counts and infiltration of tumors by type I lymphocytes with greater dimensions are associated with a good prognosis in cancers such as colon, ovarian, lung, and breast cancerCitation51–53. Generally, a low lymphocyte amount could result in a weak and insufficient immunologic reaction to the tumor. A randomized clinical trial involving more than 1000 patients revealed that, in patients receiving neoadjuvant chemotherapy, the broad infiltration of lymphocytes into tumors seemed to be more clinically beneficial than no lymphocytic infiltration, in terms of achieving a pathologic complete response after chemotherapyCitation54.

In conclusion, this study shows that elevated PLR is closely associated with poor survival outcome in patients with OC. Patients with malignant ovarian mass have higher PLR than those with benign ovarian masses. Thus, the PLR represents a predictive biomarker with great clinical utility in patients with OC.

Conflict of interest

The authors report no conflicts of interest in this work.

Source of funding

This study was supported by National Natural Science Foundation of China Grant Number [81473643], Beijing Municipal Administration of Hospitals’ Youth Programme [QML20150903], Basic-clinical cooperation Science Foundation of Capital Medical University [15JL79].

References

  • Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 2016;66:7–30
  • Jayson GC, Kohn EC, Kitchener HC, Ledermann JA. Ovarian cancer. Lancet 2014;384:1376–88
  • Cancer Research UK. 2017. Available from: http://cancerresearchuk.org
  • Doubeni CA, Doubeni AR, Myers AE. Diagnosis and management of ovarian cancer. Am Fam Physician 2016;93:937–44
  • Al Musalhi K, Al Kindi M, Al Aisary F, et al. A evaluation of HE4, CA-125, Risk of Ovarian Malignancy Algorithm (ROMA) and Risk of Malignancy Index (RMI) in the preoperative assessment of patients with adnexal mass. Oman Med J 2016;31:336–44
  • Aggarwal BB, Vijayalekshmi RV, Sung B. Targeting inflammatory pathways for prevention and therapy of cancer: short-term friend, long-term foe. Clin Cancer Res 2009;15:425–30
  • Templeton AJ, Ace O, McNamara MG, et al. Prognostic role of platelet to lymphocyte ratio in solid tumors: a systematic review and meta-analysis. Cancer Epidemiol Biomarkers Prev 2014;23:1204–12
  • Dong YW, Shi YQ, He LW, Su PZ. Prognostic significance of neutrophil-to-lymphocyte ratio in rectal cancer: a meta-analysis. Onco Targets Ther 2016;9:3127–34
  • Italiano JE, Jr., Richardson JL, Patel-Hett S, et al. Angiogenesis is regulated by a novel mechanism: pro- and antiangiogenic proteins are organized into separate platelet alpha granules and differentially released. Blood 2008;111:1227–33
  • Wouters MC, Komdeur FL, Workel HH, et al. Treatment regimen, surgical outcome, and T-cell differentiation influence prognostic benefit of tumor-infiltrating lymphocytes in high-grade serous ovarian cancer. Clin Cancer Res 2016;22:714–24
  • Khatib G, Soysal C, Cetin C, et al. Does preoperative neutrophil to lymphocyte or platelet to lymphocyte ratios have a role in predicting borderline ovarian tumors? J Ovarian Res 2016;9:78
  • Parmar MK, Torri V, Stewart L. Extracting summary statistics to perform meta-analyses of the published literature for survival endpoints. Stat Med 1998;17:2815–34
  • DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials 1986;7:177–88
  • Asher V, Lee J, Innamaa A, Bali A. Preoperative platelet lymphocyte ratio as an independent prognostic marker in ovarian cancer. Clin Transl Oncol 2011;13:499–503
  • Zhang WW, Liu KJ, Hu GL, Liang WJ. Preoperative platelet/lymphocyte ratio is a superior prognostic factor compared to other systemic inflammatory response markers in ovarian cancer patients. Tumour Biol 2015;36:8831–7
  • Miao Y, Yan Q, Li S, Li B, Feng Y. Neutrophil to lymphocyte ratio and platelet to lymphocyte ratio are predictive of chemotherapeutic response and prognosis in epithelial ovarian cancer patients treated with platinum-based chemotherapy. Cancer Biomark 2016;17:33–40
  • Raungkaewmanee S, Tangjitgamol S, Manusirivithaya S, Srijaipracharoen S, Thavaramara T. Platelet to lymphocyte ratio as a prognostic factor for epithelial ovarian cancer. J Gynecol Oncol 2012;23:265–73
  • Badora-Rybicka A, Nowara E, Starzyczny-Slota D. Neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio before chemotherapy as potential prognostic factors in patients with newly diagnosed epithelial ovarian cancer. ESMO Open 2016;1:e000039
  • Supoken A, Kleebkaow P, Chumworathayi B, Luanratanakorn S, Kietpeerakool C. Elevated preoperative platelet to lymphocyte ratio associated with decreased survival of women with ovarian clear cell carcinoma. Asian Pac J Cancer Prev 2014;15:10831–6
  • Topcu HO, Guzel AI, Ozer I, et al. Comparison of neutrophil/lymphocyte and platelet/lymphocyte ratios for predicting malignant potential of suspicious ovarian masses in gynecology practice. Asian Pac J Cancer Prev 2014;15:6239–41
  • Yildirim M, Demir Cendek B, Filiz Avsar A. Differentiation between benign and malignant ovarian masses in the preoperative period using neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios. Molec Clin Oncol 2015;3:317–21
  • Ozaksit G, Tokmak A, Kalkan H, Yesilyurt H. Value of the platelet to lymphocyte ratio in the diagnosis of ovarian neoplasms in adolescents. Asian Pac J Cancer Prev 2015;16:2037–41
  • Bakacak M, Serin S, Ercan O, et al. Utility of preoperative neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios to distinguish malignant from benign ovarian masses. J Turk Ger Gynecol Assoc 2016;17:21–5
  • Polat M, Senol T, Ozkaya E, et al. Neutrophil to lymphocyte and platelet to lymphocyte ratios increase in ovarian tumors in the presence of frank stromal invasion. Clin Transl Oncol 2016;18:457–63
  • Kemal Y, Demirag G, Ekiz K, Yucel I. Mean platelet volume could be a useful biomarker for monitoring epithelial ovarian cancer. J Obstet Gynaecol 2014;34:515–18
  • Zhou X, Du Y, Huang Z, et al. Prognostic value of PLR in various cancers: a meta-analysis. PloS One 2014;9:e101119
  • Huang CY, Yang YC, Wang KL, et al. Possible surrogate marker for an effective dose-dense chemotherapy in treating ovarian cancer. Taiwan J Obstet Gynecol 2016;55:405–9
  • Xiang J, Zhou L, Li X, et al. Preoperative monocyte-to-lymphocyte ratio in peripheral blood predicts stages, metastasis, and histological grades in patients with ovarian cancer. Transl Oncol 2017;10:33–9
  • Kokcu A, Kurtoglu E, Celik H, Tosun M, Malatyalioglu E, Ozdemir AZ. May the platelet to lymphocyte ratio be a prognostic factor for epithelial ovarian cancer? Asian Pac J Cancer Prev 2014;15:9781–4
  • Ashrafganjoei T, Mohamadianamiri M, Farzaneh F, Hosseini MS, Arab M. Investigating preoperative hematologic markers for prediction of ovarian cancer surgical outcome. Asian Pac J Cancer Prev 2016;17:1445–8
  • Qiang G, Liang C, Xiao F, et al. Prognostic significance of platelet-to-lymphocyte ratio in non-small-cell lung cancer: a meta-analysis. Onco Targets Ther 2016;9:869–76
  • Shi L, Wang L, Hou J, et al. Targeting roles of inflammatory microenvironment in lung cancer and metastasis. Cancer Metastasis Rev 2015;34:319–31
  • Alkhateeb A, Zubritsky L, Kinsman B, et al. Elevation in multiple serum inflammatory biomarkers predicts survival of pancreatic cancer patients with inoperable disease. J Gastrointest Cancer 2014;45:161–7
  • Gieger C, Radhakrishnan A, Cvejic A, et al. New gene functions in megakaryopoiesis and platelet formation. Nature 2011;480:201–208
  • Hisada Y, Geddings JE, Ay C, Mackman N. Venous thrombosis and cancer: from mouse models to clinical trials. J Thromb Haemost 2015;13:1372–82
  • Cohen JG, Tran AQ, Rimel BJ, et al. Thrombocytosis at secondary cytoreduction for recurrent ovarian cancer predicts suboptimal resection and poor survival. Gynecol Oncol 2014;132:556–9
  • Labelle M, Begum S, Hynes RO. Platelets guide the formation of early metastatic niches. Proc Natl Acad Sci USA 2014;111:E3053–61
  • Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011;473:298–307
  • Liang B, He Q, Zhong L, et al. Circulating VEGF as a biomarker for diagnosis of ovarian cancer: a systematic review and a meta-analysis. Onco Targets Ther 2015;8:1075–82
  • Lin Z, Yang Y, Huang Y, Liang J, Lu F, Lao X. Vascular endothelial growth factor receptor tyrosine kinase inhibitors versus bevacizumab in metastatic colorectal cancer: a systematic review and meta-analysis. Molec Clin Oncol 2015;3:959–67
  • Turner N, Grose R. Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer 2010;10:116–29
  • Matei D, Kelich S, Cao L, et al. PDGF BB induces VEGF secretion in ovarian cancer. Cancer Biol Ther 2007;6:1951–9
  • Grabowski J, Glode A. Ramucirumab: a vascular endothelial growth factor receptor-2 inhibitor with activity in several malignancies. Am J Health Syst Pharm 2016;73:957–68
  • Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 2006;355:2542–50
  • Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004;350:2335–42
  • Madsen CV, Steffensen KD, Olsen DA, et al. Serial measurements of serum PDGF-AA, PDGF-BB, FGF2, and VEGF in multiresistant ovarian cancer patients treated with bevacizumab. J Ovarian Res 2012;5:23
  • Economides AN, Carpenter LR, Rudge JS, et al. Cytokine traps: multi-component, high-affinity blockers of cytokine action. Nat Med 2003;9:47–52
  • Sennino B. Two is better than one: benefits of VEGF and PDGF inhibition in ovarian cancer. Cancer Biol Ther 2010;9:183–5
  • Lara PN, Jr., Douillard JY, Nakagawa K, et al. Randomized phase III placebo-controlled trial of carboplatin and paclitaxel with or without the vascular disrupting agent vadimezan (ASA404) in advanced non-small-cell lung cancer. J Clin Oncol 2011;29:2965–71
  • Hilbe W, Manegold C, Pircher A. Targeting angiogenesis in lung cancer - pitfalls in drug development. Transl Lung Cancer Res 2012;1:122–8
  • Auer K, Bachmayr-Heyda A, Sukhbaatar N, et al. Role of the immune system in the peritoneal tumor spread of high grade serous ovarian cancer. Oncotarget 2016;7:61336–54
  • Papaioannou NE, Beniata OV, Vitsos P, Tsitsilonis O, Samara P. Harnessing the immune system to improve cancer therapy. Ann Transl Med 2016;4:261
  • Stanton SE, Adams S, Disis ML. Variation in the incidence and magnitude of tumor-infiltrating lymphocytes in breast cancer subtypes: a systematic review. JAMA Oncol 2016;2:1354–60
  • Nielsen JS, Sahota RA, Milne K, et al. CD20+ tumor-infiltrating lymphocytes have an atypical CD27- memory phenotype and together with CD8+ T cells promote favorable prognosis in ovarian cancer. Clin Cancer Res 2012;18:3281–92